Intest Res.  2019 Oct;17(4):463-475. 10.5217/ir.2019.00078.

Nutritional approach as therapeutic manipulation in inflammatory bowel disease

Affiliations
  • 1Division of Gastroenterology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, Korea. htherehthere@gmail.com

Abstract

Malnutrition is observed more frequently in patients with inflammatory bowel disease (IBD) than in the general population and associated with adverse clinical outcomes. This study aimed to review the current knowledge regarding the efficacy of dietary and nutritional intervention in IBD patients. Exclusive enteral nutrition might be inferior to corticosteroid treatment in adults with active Crohn's disease (CD) but might even be superior considering the adverse effects of corticosteroid treatment in children. Total parenteral nutrition has no advantage over enteral nutrition, which is considered a more physiologic modality in organ function. Current guidelines do not yet recommend ω3-polyunsaturated fatty acid supplementation for the prevention and maintenance of remission in IBD patients. Dietary fiber supplementation could be effective in the relief of symptoms and maintenance of remission in ulcerative colitis (UC). Although vitamin D may be favorable to clinical course of IBD and bone density. Probiotic supplementation has proven to be effective in preventing and treating pouchitis for UC but is less effective in treating CD. Nutritional interventions not only correct nutritional deficiencies but also improve symptoms and clinical courses of the disease. Hence, nutritional approaches need to be developed to significantly evaluate the effectiveness of dietary interventions used to treat IBD.

Keyword

Nutrition; Crohn disease; Colitis, ulcerative; Inflammatory bowel disease

MeSH Terms

Adult
Bone Density
Child
Colitis, Ulcerative
Crohn Disease
Dietary Fiber
Enteral Nutrition
Humans
Inflammatory Bowel Diseases*
Malnutrition
Parenteral Nutrition, Total
Pouchitis
Probiotics
Vitamin D
Vitamin D

Cited by  2 articles

Evaluation of nutritional status using bioelectrical impedance analysis in patients with inflammatory bowel disease
Seung Hyuk Kim, You Sun Kim, Si Hyeong Lee, Hyun Mi Lee, Won Eui Yoon, Seo Hyun Kim, Hee Jun Myung, Jeong Seop Moon
Intest Res. 2022;20(3):321-328.    doi: 10.5217/ir.2021.00022.

Improvement of ulcerative colitis control by searching and restricting of inflammatory trigger factors in daily clinical practice
Kun-Yu Tsai, Jeng-Fu You, Tzong-Yun Tsai, Yih Jong Chern, Yu-Jen Hsu, Shu-Huan Huang, Wen-Sy Tsai
Intest Res. 2023;21(1):100-109.    doi: 10.5217/ir.2021.00110.


Reference

1. Lee D, Albenberg L, Compher C, et al. Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology. 2015; 148:1087–1106.
Article
2. Reddavide R, Rotolo O, Caruso MG, et al. The role of diet in the prevention and treatment of Inflammatory Bowel Diseases. Acta Biomed. 2018; 89(Suppl 9):60–75.
3. Sáez-González E, Mateos B, López-Muñoz P, et al. Bases for the adequate development of nutritional recommendations for patients with inflammatory bowel disease. Nutrients. 2019; 11:1062.
Article
4. Filippi J, Al-Jaouni R, Wiroth JB, Hébuterne X, Schneider SM. Nutritional deficiencies in patients with Crohn’s disease in remission. Inflamm Bowel Dis. 2006; 12:185–191.
Article
5. Vadan R, Gheorghe LS, Constantinescu A, Gheorghe C. The prevalence of malnutrition and the evolution of nutritional status in patients with moderate to severe forms of Crohn’s disease treated with Infliximab. Clin Nutr. 2011; 30:86–91.
Article
6. Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology. 2011; 140:1729–1737.
Article
7. Bernstein CN, Blanchard JF, Houston DS, Wajda A. The incidence of deep venous thrombosis and pulmonary embolism among patients with inflammatory bowel disease: a population-based cohort study. Thromb Haemost. 2001; 85:430–434.
8. Nguyen GC, Munsell M, Harris ML. Nationwide prevalence and prognostic significance of clinically diagnosable protein-calorie malnutrition in hospitalized inflammatory bowel disease patients. Inflamm Bowel Dis. 2008; 14:1105–1111.
9. Card T, Hubbard R, Logan RF. Mortality in inflammatory bowel disease: a population-based cohort study. Gastroenterology. 2003; 125:1583–1590.
Article
10. Ashton JJ, Gavin J, Beattie RM. Exclusive enteral nutrition in Crohn’s disease: evidence and practicalities. Clin Nutr. 2019; 38:80–89.
11. de Jong NS, Leach ST, Day AS. Polymeric formula has direct anti-inflammatory effects on enterocytes in an in vitro model of intestinal inflammation. Dig Dis Sci. 2007; 52:2029–2036.
Article
12. Fell JM. Control of systemic and local inflammation with transforming growth factor beta containing formulas. JPEN J Parenter Enteral Nutr. 2005; 29(4 Suppl):S126–S128.
13. Grover Z, Muir R, Lewindon P. Exclusive enteral nutrition induces early clinical, mucosal and transmural remission in paediatric Crohn’s disease. J Gastroenterol. 2014; 49:638–645.
Article
14. Sanderson IR, Boulton P, Menzies I, Walker-Smith JA. Improvement of abnormal lactulose/rhamnose permeability in active Crohn’s disease of the small bowel by an elemental diet. Gut. 1987; 28:1073–1076.
Article
15. Levine A, Wine E. Effects of enteral nutrition on Crohn’s disease: clues to the impact of diet on disease pathogenesis. Inflamm Bowel Dis. 2013; 19:1322–1329.
16. Kaakoush NO, Day AS, Leach ST, Lemberg DA, Mitchell HM. Reduction in gut microbial diversity as a mechanism of action of exclusive enteral nutrition. Am J Gastroenterol. 2016; 111:1033.
Article
17. Gerasimidis K, Bertz M, Hanske L, et al. Decline in presumptively protective gut bacterial species and metabolites are paradoxically associated with disease improvement in pediatric Crohn’s disease during enteral nutrition. Inflamm Bowel Dis. 2014; 20:861–871.
Article
18. Narula N, Dhillon A, Zhang D, Sherlock ME, Tondeur M, Zachos M. Enteral nutritional therapy for induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 2018; 4:CD 000542.
19. Swaminath A, Feathers A, Ananthakrishnan AN, Falzon L, Li Ferry S. Systematic review with meta-analysis: enteral nutrition therapy for the induction of remission in paediatric Crohn’s disease. Aliment Pharmacol Ther. 2017; 46:645–656.
20. Fell JM, Paintin M, Arnaud-Battandier F, et al. Mucosal healing and a fall in mucosal pro-inflammatory cytokine mRNA induced by a specific oral polymeric diet in paediatric Crohn’s disease. Aliment Pharmacol Ther. 2000; 14:281–289.
Article
21. Grover Z, Burgess C, Muir R, Reilly C, Lewindon PJ. Early mucosal healing with exclusive enteral nutrition is associated with improved outcomes in newly diagnosed children with luminal Crohn’s disease. J Crohns Colitis. 2016; 10:1159–1164.
Article
22. Bannerjee K, Camacho-Hübner C, Babinska K, et al. Anti-inflammatory and growth-stimulating effects precede nutritional restitution during enteral feeding in Crohn disease. J Pediatr Gastroenterol Nutr. 2004; 38:270–275.
23. Berni Canani R, Terrin G, Borrelli O, et al. Short- and long-term therapeutic efficacy of nutritional therapy and corticosteroids in paediatric Crohn’s disease. Dig Liver Dis. 2006; 38:381–387.
Article
24. Gerasimidis K, Talwar D, Duncan A, et al. Impact of exclusive enteral nutrition on body composition and circulating micronutrients in plasma and erythrocytes of children with active Crohn’s disease. Inflamm Bowel Dis. 2012; 18:1672–1681.
Article
25. Gerasimidis K, Barclay A, Papangelou A, et al. The epidemiology of anemia in pediatric inflammatory bowel disease: prevalence and associated factors at diagnosis and follow-up and the impact of exclusive enteral nutrition. Inflamm Bowel Dis. 2013; 19:2411–2422.
26. Grover Z, Lewindon P. Two-year outcomes after exclusive enteral nutrition induction are superior to corticosteroids in pediatric Crohn’s disease treated early with thiopurines. Dig Dis Sci. 2015; 60:3069–3074.
Article
27. Connors J, Basseri S, Grant A, et al. Exclusive enteral nutrition therapy in paediatric Crohn’s disease results in long-term avoidance of corticosteroids: results of a propensity-score matched cohort analysis. J Crohns Colitis. 2017; 11:1063–1070.
Article
28. Cohen-Dolev N, Sladek M, Hussey S, et al. Differences in outcomes over time with exclusive enteral nutrition compared with steroids in children with mild to moderate Crohn’s disease: results from the GROWTH CD Study. J Crohns Colitis. 2018; 12:306–312.
Article
29. Ruemmele FM, Veres G, Kolho KL, et al. Consensus guidelines of ECCO/ESPGHAN on the medical management of pediatric Crohn’s disease. J Crohns Colitis. 2014; 8:1179–1207.
30. Forbes A, Escher J, Hébuterne X, et al. ESPEN guideline: clinical nutrition in inflammatory bowel disease. Clin Nutr. 2017; 36:321–347.
Article
31. González-Huix F, Fernández-Bañares F, Esteve-Comas M, et al. Enteral versus parenteral nutrition as adjunct therapy in acute ulcerative colitis. Am J Gastroenterol. 1993; 88:227–232.
32. Shivashankar R, Lewis JD. The role of diet in inflammatory bowel disease. Curr Gastroenterol Rep. 2017; 19:22.
Article
33. Green N, Miller T, Suskind D, Lee D. A review of dietary therapy for IBD and a vision for the future. Nutrients. 2019; 11:947.
Article
34. Okada M, Yao T, Yamamoto T, et al. Controlled trial comparing an elemental diet with prednisolone in the treatment of active Crohn’s disease. Hepatogastroenterology. 1990; 37:72–80.
35. Teahon K, Bjarnason I, Pearson M, Levi AJ. Ten years’ experience with an elemental diet in the management of Crohn’s disease. Gut. 1990; 31:1133–1137.
Article
36. Akobeng AK, Zhang D, Gordon M, MacDonald JK. Enteral nutrition for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2018; 8:CD005984.
Article
37. Nguyen DL, Palmer LB, Nguyen ET, McClave SA, Martindale RG, Bechtold ML. Specialized enteral nutrition therapy in Crohn’s disease patients on maintenance infliximab therapy: a meta-analysis. Therap Adv Gastroenterol. 2015; 8:168–175.
38. Yang Q, Gao X, Chen H, et al. Efficacy of exclusive enteral nutrition in complicated Crohn’s disease. Scand J Gastroenterol. 2017; 52:995–1001.
Article
39. Hu D, Ren J, Wang G, et al. Exclusive enteral nutritional therapy can relieve inflammatory bowel stricture in Crohn’s disease. J Clin Gastroenterol. 2014; 48:790–795.
40. Heerasing N, Thompson B, Hendy P, et al. Exclusive enteral nutrition provides an effective bridge to safer interval elective surgery for adults with Crohn’s disease. Aliment Pharmacol Ther. 2017; 45:660–669.
Article
41. Zheng XB, Peng X, Xie XY, et al. Enteral nutrition is associated with a decreased risk of surgical intervention in Crohn’s disease patients with spontaneous intra-abdominal abscess. Rev Esp Enferm Dig. 2017; 109:834–842.
Article
42. Scolapio JS. The role of total parenteral nutrition in the management of patients with acute attacks of inflammatory bowel disease. J Clin Gastroentero. 1999; 29:223–224.
Article
43. Dickinson RJ, Ashton MG, Axon AT, Smith RC, Yeung CK, Hill GL. Controlled trial of intravenous hyperalimentation and total bowel rest as an adjunct to the routine therapy of acute colitis. Gastroenterology. 1980; 79:1199–1204.
44. Müller JM, Keller HW, Erasmi H, Pichlmaier H. Total parenteral nutrition as the sole therapy in Crohn’s disease: a prospective study. Br J Surg. 1983; 70:40–43.
Article
45. Ostro MJ, Greenberg GR, Jeejeebhoy KN. Total parenteral nutrition and complete bowel rest in the management of Crohn’s disease. JPEN J Parenter Enteral Nutr. 1985; 9:280–287.
Article
46. McIntyre PB, Powell-Tuck J, Wood SR, et al. Controlled trial of bowel rest in the treatment of severe acute colitis. Gut. 1986; 27:481–485.
Article
47. Greenberg GR, Fleming CR, Jeejeebhoy KN, Rosenberg IH, Sales D, Tremaine WJ. Controlled trial of bowel rest and nutritional support in the management of Crohn’s disease. Gut. 1988; 29:1309–1315.
Article
48. Richman E, Rhodes JM. Review article: evidence-based dietary advice for patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2013; 38:1156–1171.
49. Triantafillidis JK, Papalois AE. The role of total parenteral nutrition in inflammatory bowel disease: current aspects. Scand J Gastroenterol. 2014; 49:3–14.
Article
50. Egberg MD, Galanko JA, Barnes EL, Kappelman MD. Thrombotic and infectious risks of parenteral nutrition in hospitalized pediatric inflammatory bowel disease. Inflamm Bowel Dis. 2019; 25:601–609.
Article
51. Gouma DJ, von Meyenfeldt MF, Rouflart M, Soeters PB. Preoperative total parenteral nutrition (TPN) in severe Crohn’s disease. Surgery. 1988; 103:648–652.
52. Han PD, Burke A, Baldassano RN, Rombeau JL, Lichtenstein GR. Nutrition and inflammatory bowel disease. Gastroenterol Clin North Am. 1999; 28:423–443.
Article
53. Lashner BA, Evans AA, Hanauer SB. Preoperative total parenteral nutrition for bowel resection in Crohn’s disease. Dig Dis Sci. 1989; 34:741–746.
Article
54. Duvall MG, Levy BD. DHA- and EPA-derived resolvins, protectins, and maresins in airway inflammation. Eur J Pharmacol. 2016; 785:144–155.
Article
55. Baker EJ, Miles EA, Burdge GC, Yaqoob P, Calder PC. Metabolism and functional effects of plant-derived omega-3 fatty acids in humans. Prog Lipid Res. 2016; 64:30–56.
Article
56. Martindale RG, Warren MM, McClave SA. Does the use of specialized proresolving molecules in critical care offer a more focused approach to controlling inflammation than that of fish oils? Curr Opin Clin Nutr Metab Care. 2016; 19:151–154.
57. Mbodji K, Charpentier C, Guérin C, et al. Adjunct therapy of n-3 fatty acids to 5-ASA ameliorates inflammatory score and decreases NF-kappaB in rats with TNBS-induced colitis. J Nutr Biochem. 2013; 24:700–705.
58. Nieto N, Torres MI, Ríos A, Gil A. Dietary polyunsaturated fatty acids improve histological and biochemical alterations in rats with experimental ulcerative colitis. J Nutr. 2002; 132:11–19.
59. Borniquel S, Jädert C, Lundberg JO. Dietary conjugated linoleic acid activates PPARgamma and the intestinal trefoil factor in SW480 cells and mice with dextran sulfate sodium-induced colitis. J Nutr. 2012; 142:2135–2140.
60. Arita M, Yoshida M, Hong S, et al. Resolvin E1, an endogenous lipid mediator derived from omega-3 eicosapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Proc Natl Acad Sci U S A. 2005; 102:7671–7676.
Article
61. Ibrahim A, Aziz M, Hassan A, et al. Dietary alpha-linolenic acid-rich formula reduces adhesion molecules in rats with experimental colitis. Nutrition. 2012; 28:799–802.
62. Tyagi A, Kumar U, Santosh VS, Reddy S, Mohammed SB, Ibrahim A. Partial replacement of dietary linoleic acid with long chain n-3 polyunsaturated fatty acids protects against dextran sulfate sodium-induced colitis in rats. Prostaglandins Leukot Essent Fatty Acids. 2014; 91:289–297.
Article
63. IBD in EPIC Study Investigators, Tjonneland A, Overvad K, et al. Linoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: a nested case-control study within a European prospective cohort study. Gut. 2009; 58:1606–1611.
Article
64. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. Long-term intake of dietary fat and risk of ulcerative colitis and Crohn’s disease. Gut. 2014; 63:776–784.
65. Ananthakrishnan AN, Khalili H, Song M, et al. High school diet and risk of Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis. 2015; 21:2311–2319.
Article
66. Maconi G, Ardizzone S, Cucino C, Bezzio C, Russo AG, Bianchi Porro G. Pre-illness changes in dietary habits and diet as a risk factor for inflammatory bowel disease: a case-control study. World J Gastroenterol. 2010; 16:4297–4304.
Article
67. Reif S, Klein I, Lubin F, Farbstein M, Hallak A, Gilat T. Pre-illness dietary factors in inflammatory bowel disease. Gut. 1997; 40:754–760.
Article
68. John S, Luben R, Shrestha SS, Welch A, Khaw KT, Hart AR. Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective cohort study. Eur J Gastroenterol Hepatol. 2010; 22:602–606.
69. Mozaffari H, Daneshzad E, Larijani B, Bellissimo N, Azadbakht L. Dietary intake of fish, n-3 polyunsaturated fatty acids, and risk of inflammatory bowel disease: a systematic review and meta-analysis of observational studies. Eur J Nutr. [published online ahead of print January 24, 2019]. https://doi.org/10.1007/s00394-019-01901-0.
Article
70. Feagan BG, Sandborn WJ, Mittmann U, et al. Omega-3 free fatty acids for the maintenance of remission in Crohn disease: the EPIC randomized controlled trials. JAMA. 2008; 299:1690–1697.
Article
71. Lev-Tzion R, Griffiths AM, Leder O, Turner D. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2014; (2):CD006320.
Article
72. Turner D, Shah PS, Steinhart AH, Zlotkin S, Griffiths AM. Maintenance of remission in inflammatory bowel disease using omega-3 fatty acids (fish oil): a systematic review and meta-analyses. Inflamm Bowel Dis. 2011; 17:336–345.
Article
73. Turner D, Steinhart AH, Griffiths AM. Omega 3 fatty acids (fish oil) for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2007; (3):CD006443.
74. Uchiyama K, Nakamura M, Odahara S, et al. N-3 polyunsaturated fatty acid diet therapy for patients with inflammatory bowel disease. Inflamm Bowel Dis. 2010; 16:1696–1707.
75. Grimstad T, Berge RK, Bohov P, et al. Salmon diet in patients with active ulcerative colitis reduced the simple clinical colitis activity index and increased the anti-inflammatory fatty acid index: a pilot study. Scand J Clin Lab Invest. 2011; 71:68–73.
76. Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011; 3:858–876.
77. Kim S, Kim JH, Park BO, Kwak YS. Perspectives on the therapeutic potential of short-chain fatty acid receptors. BMB Rep. 2014; 47:173–178.
78. Singh N, Gurav A, Sivaprakasam S, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014; 40:128–139.
79. Saeedi BJ, Kao DJ, Kitzenberg DA, et al. HIF-dependent regulation of claudin-1 is central to intestinal epithelial tight junction integrity. Mol Biol Cell. 2015; 26:2252–2262.
80. Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. 2017; 8:172–184.
81. Huda-Faujan N, Abdulamir AS, Fatimah AB, et al. The impact of the level of the intestinal short chain fatty acids in inflammatory bowel disease patients versus healthy subjects. Open Biochem J. 2010; 4:53–58.
82. Hague A, Singh B, Paraskeva C. Butyrate acts as a survival factor for colonic epithelial cells: further fuel for the in vivo versus in vitro debate. Gastroenterology. 1997; 112:1036–1040.
83. Benjamin JL, Hedin CR, Koutsoumpas A, et al. Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn’s disease. Gut. 2011; 60:923–929.
84. Fernández-Bañares F, Hinojosa J, Sánchez-Lombraña JL, et al. Randomized clinical trial of Plantago ovata seeds (dietary fiber) as compared with mesalamine in maintaining remission in ulcerative colitis. Spanish Group for the Study of Crohn’s Disease and Ulcerative Colitis (GETECCU). Am J Gastroenterol. 1999; 94:427–433.
85. Wedlake L, Slack N, Andreyev HJ, Whelan K. Fiber in the treatment and maintenance of inflammatory bowel disease: a systematic review of randomized controlled trials. Inflamm Bowel Dis. 2014; 20:576–586.
86. Sugihara K, Morhardt TL, Kamada N. The role of dietary nutrients in inflammatory bowel disease. Front Immunol. 2019; 9:3183.
87. Bartel G, Weiss I, Turetschek K, et al. Ingested matter affects intestinal lesions in Crohn’s disease. Inflamm Bowel Dis. 2008; 14:374–382.
88. Morgan XC, Tickle TL, Sokol H, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012; 13:R79.
89. Davenport M, Poles J, Leung JM, et al. Metabolic alterations to the mucosal microbiota in inflammatory bowel disease. Inflamm Bowel Dis. 2014; 20:723–731.
90. Kovatcheva-Datchary P, Nilsson A, Akrami R, et al. Dietary fiber-induced improvement in glucose metabolism is associated with increased abundance of prevotella. Cell Metab. 2015; 22:971–982.
91. Silveira ALM, Ferreira AVM, de Oliveira MC, et al. Preventive rather than therapeutic treatment with high fiber diet attenuates clinical and inflammatory markers of acute and chronic DSS-induced colitis in mice. Eur J Nutr. 2017; 56:179–191.
92. Vanhauwaert E, Matthys C, Verdonck L, De Preter V. Low-residue and low-fiber diets in gastrointestinal disease management. Adv Nutr. 2015; 6:820–827.
93. Zhang YJ, Li S, Gan RY, Zhou T, Xu DP, Li HB. Impacts of gut bacteria on human health and diseases. Int J Mol Sci. 2015; 16:7493–7519.
94. Levenstein S, Prantera C, Luzi C, D’Ubaldi A. Low residue or normal diet in Crohn’s disease: a prospective controlled study in Italian patients. Gut. 1985; 26:989–993.
95. Brotherton CS, Taylor AG, Bourguignon C, Anderson JG. A high-fiber diet may improve bowel function and health-related quality of life in patients with Crohn disease. Gastroenterol Nurs. 2014; 37:206–216.
96. Chiba M, Tsuji T, Nakane K, Komatsu M. High amount of dietary fiber not harmful but favorable for Crohn disease. Perm J. 2015; 19:58–61.
97. Hwang C, Ross V, Mahadevan U. Popular exclusionary diets for inflammatory bowel disease: the search for a dietary culprit. Inflamm Bowel Dis. 2014; 20:732–741.
98. Fletcher J. Vitamin D deficiency in patients with inflammatory bowel disease. Br J Nurs. 2016; 25:846–851.
99. Nielsen OH, Rejnmark L, Moss AC. Role of vitamin D in the natural history of inflammatory bowel disease. J Crohns Colitis. 2018; 12:742–752.
100. Fletcher J, Cooper SC, Ghosh S, Hewison M. The role of vitamin D in inflammatory bowel disease: mechanism to management. Nutrients. 2019; 11:1019.
101. Ghosh S, Cowen S, Hannan WJ, Ferguson A. Low bone mineral density in Crohn’s disease, but not in ulcerative colitis, at diagnosis. Gastroenterology. 1994; 107:1031–1039.
102. Schäffler H, Schmidt M, Huth A, Reiner J, Glass Ä, Lamprecht G. Clinical factors are associated with vitamin D levels in IBD patients: a retrospective analysis. J Dig Dis. 2018; 19:24–32.
103. Liu PT, Stenger S, Li H, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006; 311:1770–1773.
104. Ananthakrishnan AN. Vitamin D and inflammatory bowel disease. Gastroenterol Hepatol (N Y). 2016; 12:513–515.
105. Zhao H, Zhang H, Wu H, et al. Protective role of 1,25(OH)2 vitamin D3 in the mucosal injury and epithelial barrier disruption in DSS-induced acute colitis in mice. BMC Gastroenterol. 2012; 12:57.
106. Ananthakrishnan AN, Khalili H, Higuchi LM, et al. Higher predicted vitamin D status is associated with reduced risk of Crohn’s disease. Gastroenterology. 2012; 142:482–489.
107. Rosen CJ. Clinical practice: vitamin D insufficiency. N Engl J Med. 2011; 364:248–254.
108. Ananthakrishnan AN, Cagan A, Gainer VS, et al. Normalization of plasma 25-hydroxy vitamin D is associated with reduced risk of surgery in Crohn’s disease. Inflamm Bowel Dis. 2013; 19:1921–1927.
109. Li J, Chen N, Wang D, Zhang J, Gong X. Efficacy of vitamin D in treatment of inflammatory bowel disease: a meta-analysis. Medicine (Baltimore). 2018; 97:e12662.
110. Garg M, Hendy P, Ding JN, Shaw S, Hold G, Hart A. The effect of vitamin D on intestinal inflammation and faecal microbiota in patients with ulcerative colitis. J Crohns Colitis. 2018; 12:963–972.
111. Zator ZA, Cantu SM, Konijeti GG, et al. Pretreatment 25-hydroxyvitamin D levels and durability of anti-tumor necrosis factor-α therapy in inflammatory bowel diseases. JPEN J Parenter Enteral Nutr. 2014; 38:385–391.
112. Winter RW, Collins E, Cao B, Carrellas M, Crowell AM, Korzenik JR. Higher 25-hydroxyvitamin D levels are associated with greater odds of remission with anti-tumour necrosis factor-alpha medications among patients with inflammatory bowel diseases. Aliment Pharmacol Ther. 2017; 45:653–659.
113. Ananthakrishnan AN, Cheng SC, Cai T, et al. Association between reduced plasma 25-hydroxy vitamin D and increased risk of cancer in patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol. 2014; 12:821–827.
114. Kabbani TA, Koutroubakis IE, Schoen RE, et al. Association of vitamin D level with clinical status in inflammatory bowel disease: a 5-year longitudinal study. Am J Gastroenterol. 2016; 111:712–719.
115. Sharifi A, Hosseinzadeh-Attar MJ, Vahedi H, Nedjat S. A randomized controlled trial on the effect of vitamin D3 on inflammation and cathelicidin gene expression in ulcerative colitis patients. Saudi J Gastroenterol. 2016; 22:316–323.
116. Hlavaty T, Krajcovicova A, Payer J. Vitamin D therapy in inflammatory bowel diseases: who, in what form, and how much? J Crohns Colitis. 2015; 9:198–209.
117. Lichtenstein L, Avni-Biron I, Ben-Bassat O. Probiotics and prebiotics in Crohn’s disease therapies. Best Pract Res Clin Gastroenterol. 2016; 30:81–88.
118. Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology. 2008; 134:577–594.
119. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018; 11:1–10.
120. Ng SC, Hart AL, Kamm MA, Stagg AJ, Knight SC. Mechanisms of action of probiotics: recent advances. Inflamm Bowel Dis. 2009; 15:300–310.
121. Bindels LB, Delzenne NM, Cani PD, Walter J. Towards a more comprehensive concept for prebiotics. Nat Rev Gastroenterol Hepatol. 2015; 12:303–310.
122. Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces boulardii in maintenance treatment of Crohn’s disease. Dig Dis Sci. 2000; 45:1462–1464.
123. Derwa Y, Gracie DJ, Hamlin PJ, Ford AC. Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease. Aliment Pharmacol Ther. 2017; 46:389–400.
124. Ganji-Arjenaki M, Rafieian-Kopaei M. Probiotics are a good choice in remission of inflammatory bowel diseases: a meta analysis and systematic review. J Cell Physiol. 2018; 233:2091–2103.
125. Isaacs K, Herfarth H. Role of probiotic therapy in IBD. Inflamm Bowel Dis. 2008; 14:1597–1605.
126. Kruis W, Fric P, Pokrotnieks J, et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut. 2004; 53:1617–1623.
127. Nguyen N, Zhang B, Holubar SD, Pardi DS, Singh S. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst Rev. 2019; 5:CD001176.
128. Mimura T, Rizzello F, Helwig U, et al. Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut. 2004; 53:108–114.
129. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind, placebo-controlled trial. Gastroenterology. 2003; 124:1202–1209.
130. Gosselink MP, Schouten WR, van Lieshout LM, Hop WC, Laman JD, Ruseler-van Embden JG. Delay of the first onset of pouchitis by oral intake of the probiotic strain Lactobacillus rhamnosus GG. Dis Colon Rectum. 2004; 47:876–884.
Full Text Links
  • IR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr